US20180312600A1 - Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer - Google Patents

Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer Download PDF

Info

Publication number
US20180312600A1
US20180312600A1 US15/769,689 US201615769689A US2018312600A1 US 20180312600 A1 US20180312600 A1 US 20180312600A1 US 201615769689 A US201615769689 A US 201615769689A US 2018312600 A1 US2018312600 A1 US 2018312600A1
Authority
US
United States
Prior art keywords
sirpa
compound
macrophages
inflammatory
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/769,689
Other languages
English (en)
Inventor
Nicolas Poirier
Bernard Vanhove
Vanessa Gauttier
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
OSE Immunotherapeutics SA
Original Assignee
OSE Immunotherapeutics SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by OSE Immunotherapeutics SA filed Critical OSE Immunotherapeutics SA
Assigned to OSE IMMUNOTHERAPEUTICS reassignment OSE IMMUNOTHERAPEUTICS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GAUTTIER, Vanessa, POIRIER, NICOLAS, VANHOVE, BERNARD
Publication of US20180312600A1 publication Critical patent/US20180312600A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0645Macrophages, e.g. Kuepfer cells in the liver; Monocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy

Definitions

  • the present invention pertains to the field of immunotherapy. More specifically, the present invention provides a method for inhibiting M2-type macrophages polarization in order to induce a pro-inflammatory environment and consequently allows appropriate immune responses in cancers, infectious diseases, vaccination, trauma and chronic inflammatory diseases.
  • the present invention concerns in particular the use of an anti-SIRPa compound able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophages.
  • an anti-SIRPa compound able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophages.
  • such compound is used to treat cancer.
  • this invention allows to treat cancer through an indirect pathway involving the immune system.
  • New generation immunotherapeutic molecules revolutionizing cancer treatments, block suppressor mechanisms of these cells, allowing the effector T cells (Teff) to exercise their action.
  • This is the concept called “Inhibit inhibitors”.
  • An anti-CTLA-4 antibody (Yervoy®) has been the first molecule for treating metastatic forms of malignant melanoma prolonging the mean survival of patients of 6 to 10 months, with a quarter of patients still alive after 2 years. Unfortunately, these results, as spectacular as they are, still do not cure most patients.
  • the present invention relies on an “inhibit inhibitors” approach and provides a new method useful in immunotherapy. More specifically, the present invention pertains to a method for modifying macrophage polarization in order to induce a pro-inflammatory environment.
  • the method consists in the use of an anti-SIRPa compound able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophages, for inhibiting the anti-inflammatory signal provided by M2-type macrophages and favoring the pro-inflammatory signal provided by M1-type macrophages.
  • This approach allows to reestablish an inflammatory environment favorable to the action of the T effector cells, in particular in eliminating the cancer cells.
  • Macrophages are cells that have the highest plasticity of the hematopoietic system. They are involved both in innate immunity (phagocytosis capacity) and in adaptive immunity (cell polarization), but also in ontogeny, in homeostasis and in tissue repair (Mantovani et al., 2013; Wynn et al., 2013). Macrophages are present in all tissues. They have a large phenotypic and functional diversity. During ontogeny, these cells also exhibit a diversity of origins which persists into adulthood. In the tissues, monocytes-macrophages respond to environmental stimuli (product from microbial infection, damaged cells, activated lymphocytes) and acquire distinct phenotypes. For a long time, these cells have been classified according to their function in a binary manner in connection with the inflammatory condition.
  • Macrophages are categorized simplistically into 2 sub-populations or states of polarization (or activation): classical activation phenotype M1 and the alternative activation phenotype M2 (Gabrilovich et al., 2012).
  • the M1 classification is associated in vitro with the use of IFNg factor alone or in combination with microbial factors such as LPS or inflammatory cytokines such as TNF- ⁇ and GM-CSF.
  • the polarization M2 is rather associated with the IL4 or IL13 (Stein et al., 1992).
  • cytokines are also identified as inducing M2 type polarization such as IL33, which induces overexpression of Arg1 (arginase 1), CCL24 or CCL17 playing a role in inflammation.
  • IL21 and more commonly CSF1 are major players in the polarization of macrophages. Macrophages may also acquire the status of “M2-like”, sharing common characteristics of M2.
  • stimuli such as immune complexes associated with LPS, IL-1, glucocorticoids, TGF beta, Wnt5a and IL10 result in a functional phenotype of type “M2-like”.
  • M1, M2 and M2-like macrophages represent only the extremes on a continuum of functional states that must be integrated in an environmental complex system.
  • M1 macrophages present IL12 high IL23 high and IL10 low phenotype and produce molecular effectors such as reactive oxygen species (ROS) and intermediates of Nitric Oxide (NO) and inflammatory cytokines (IL1b, TNF- ⁇ , IL-6).
  • ROS reactive oxygen species
  • NO Nitric Oxide
  • IL1b IL1b
  • TNF- ⁇ IL-6
  • M1 macrophages participate in Th1 responses, play a role in resistance against intracellular parasites and are key effectors in the elimination of tumor cells.
  • M2 macrophages have an IL12 low , IL23 low and IL10 high phenotype with a variability in the production of inflammatory cytokines according to stimuli present in the environment.
  • M2 cells display on their surface a strong expression of scavenger, mannose and galactose-type receptors.
  • the metabolism of arginine is changed to an ornithine and polyamines metabolism.
  • M2 macrophages are generally associated to a Th2 type response, to a parasite clearance, to a decrease of inflammation, to tissue repair promotion, angiogenesis, tumor growth and immune regulation.
  • M1 and M2 also have distinct expression profiles of chemokines.
  • M1 macrophages express CXCL9 and CXCL10 chemokines which are known for attracting Th1, while M2 macrophages express CCL17, CCL22 and CCL24.
  • Chemokines such as CCL2 and CXCL4 can also polarize macrophages to an M2-like phenotype.
  • macrophages Depending on their polarization state, macrophages have different characteristics in terms of iron, folate and glucose metabolisms.
  • M1 express large amounts of proteins involved in iron storage, such as Ferritin, while they express only weakly Ferroportin, involved in the iron exportation to the extracellular medium.
  • M2 macrophages express low levels of Ferritin but high levels of Ferroportin. This difference can result in functional outcomes, such as a bacteriostatic effect of M1 (protection against infection) and an effect promoting tissue repair by M2 macrophages, which also promote the tumor growth, as observed in some studies.
  • the management of iron by macrophages according to their polarity is an important element underlining the importance of controlling the polarization of macrophages according to the condition of an individual.
  • macrophages face an oxygen gradient in tissues under normal or pathological conditions. Macrophages or monocytes adapt to this gradient by modifying their glycolytic metabolism.
  • the HIF1 and 2 are transcriptional factors leaders of these changes, including expression of chemokines or chemokine receptor CXCR4 or CXCL12 and VEGF (an angiogenic factor). Macrophages are involved in the tissue response to hypoxic conditions.
  • polyamines in the cell environment appears to be a type 2 macrophage polarization factor.
  • the present invention aims to modulate the polarization of macrophages in order to inhibit the anti-inflammatory M2-type macrophages and/or to favor the pro-inflammatory M1-type macrophages.
  • the adaptive immune response is suppressed/blocked by activating a number of pathways leading to the inhibition of differentiation and activation of dendritic cells (via the presence in tumor microenvironment factors such as IL10 and VEGF).
  • tumor microenvironment factors such as IL10 and VEGF.
  • Treg regulatory T cells
  • tumor suppressor cells such as MDSC (myeloid derived suppressor cells) and TAM (Tumor Associated Macrophages or M2), affects tumor development of bad prognosis by the secretion of cytokines, growth factors, enzymes degrading the extracellular matrix and proteases (Cornelissen et al., 2012).
  • Immunotherapy is safe but in some cancer has a moderate efficacy partly due to the presence of immunosuppressive cells in peripheral blood, lymphoid organs and within the tumour environment that hamper immunotherapeutic treatments.
  • Several strategies have been performed or are currently tested to improve the efficacy of immunotherapy by acting on suppressive cells such as MDSCs, Tregs and TAMs, which are increased in most cancer patients. It is becoming increasingly clear that these populations contribute to the impaired antitumour responses frequently observed in cancer patients.
  • the present invention aims to modify the M1/M2 balance of macrophage population to favor the M1-type macrophages, in order to provide an immune environment propitious to immunotherapy.
  • Macrophages are found in large numbers in tumors. Originally it was thought that this cell population was involved in an anti-tumor response but many experimental and clinical studies have shown that macrophages are involved in the tumor initiation and progression as well as in the metastatic process. During the tumor process, macrophages secrete pro-inflammatory cytokines such as IFN ⁇ , TNF- ⁇ and IL6, attracting other immune cells creating chronic inflammation and causing the initiation and tumor progression.
  • tumor macrophages adopt an immunosuppressive cellular profile and are less active, allowing tumor growth and transition to malignancy. TAMs are responsible for migration, extravasation and invasion of tumor cells (metastasis) and are involved in tumor angiogenesis (Qian and Pollard, 2010; DeNardo et al., 2010; Hanahan and Coussens, 2012).
  • MHCII MHC class II
  • VCAM VCAM
  • CD11c MHC class II
  • TAM's function was previously based on anti-tumor M1 macrophage type (iNOS inducible) and M2 pro-tumoral ARG-positive macrophage type.
  • the present invention pertains to the inhibition of TAM in order to decrease or prevent the tumoral process, including the metastatic process.
  • SIRPa Signal regulatory protein alpha
  • CD172a or SHPS-1 and herein noted “SIRPa” Signal regulatory protein alpha
  • SIRPa was first identified as a membrane protein mainly present on macrophages and myeloid cells that was associated with the Src homology region 2 (SH2) domain—containing phosphatases—SHP-1 and SHP-2.
  • SIRPa is the prototypic member of the SIRP paired receptor family of closely related SIRP proteins. Engagement of SIRPa by CD47 provides a downregulatory signal that inhibits host cell phagocytosis, and CD47 therefore functions as a “don't-eat-me” signal.
  • SIRPa is expressed on monocytes, most subpopulations of tissue macrophages, granulocytes, subsets of dendritic cells (DCs) in tissues, some bone marrow progenitor cells, and to varying levels on neurons, with a notably high expression in synapse-rich areas of the brain, such as the granular layer of the cerebellum and the hippocampus (Seiffert et al, 1994; Adams et al, 1998; Milling et al, 2010).
  • DCs dendritic cells
  • the SIRPa interaction with CD47 is largely described and provides a downregulatory signal that inhibits host cell phagocytosis (see review Barclay et al, Annu. Rev. Immunol., 2014). Both CD47 and SIRPa also engage in other interactions. Investigators have suggested that the lung surfactant proteins SP-A and SP-D control inflammatory responses in the lung through interactions with SIRPa (Janssen et al., 2008).
  • CD47-SIRPa interactions One of the best characterized physiological functions of CD47-SIRPa interactions is their role in the homeostasis of hematopoietic cells, in particular red blood cells and platelets. Because CD47 acts as a don't-eat-me signal and, as such, is an important determinant of host cell phagocytosis by macrophages, the potential contribution of CD47-SIRPa interactions in cancer cell clearance has been intensely investigated in recent years.
  • SIRPa/CD47 pathway is nowadays also subject to different pharmaceutical developments, all directed towards enhancement of macrophages phagocytosis.
  • cancer cells carry aberrant cargo such as unfamiliar proteins or normal proteins at abnormal levels, yet these cells frequently subvert innate immune control mechanisms by concurrently overexpressing immunoregulatory molecules.
  • CD47 Barclay and Van den Berg, 2014
  • CD47 has interactions with several different ligands such as SIRPa.
  • Blockade of the CD47/SIRPa pathway by enhancing antibody-dependent phagocytosis by macrophages, has been described to synergize with depleting therapeutic anticancer antibodies (Weiskopf et al., 2013) such as Trastuzumab (anti-Her2), Cetuximab (anti-EGFR), Rituximab (anti-CD20) and Alemtuzumab (anti-CD52).
  • SIRPa has been described to be implicated into the phagocytic function of myeloid cells, the antigen presentation and cytokine secretion of dendritic cells and trafficking of mature granulocytes.
  • the function of SIRPa on macrophage polarization and their potent suppressive function during tumorigenesis have never been described.
  • WO2010/130053 disclosed a method for treating hematological cancer comprising modulating the interaction between human SIRPa and CD47.
  • This document showed that the blockade of SIRPa-CD47 induces the activation of the innate immune system via the phagocytosis pathway.
  • myeloid cells were used and the transplant was rejected when animals were treated with an antagonist of CD47. This result suggests an increase of phagocytosis upon treatment with anti-CD47, but not a modification in the polarization state of the macrophages nor a modification into a pro-inflammatory function of the macrophages.
  • a method for inhibiting cell functioning for use in anti-inflammatory and anti-tumor therapies was described in WO0066159.
  • This method comprises administering a drug comprising a substance that specifically recognizes the extracellular domain of SIRPa and that inhibits the functioning of pathologic myeloid cells.
  • the inventors of this patent application claim that an anti-SIRPa antibody specific to the extracellular domain has the property to block the inflammation and inhibit macrophage phagocytosis (referred to as the functioning of pathologic myeloid cells in the text). This effect is in total contradiction with the results disclosed below and do not suggest any effect of an anti-SIRPa antibody on the macrophage polarization nor on pro-inflammatory function.
  • a CD47-Fc and a CD47-extended fusobody molecules that bind to SIRPa were studied and claimed in the patent application number WO2012/172521. These molecules have been claimed to be able to inhibit immune complex-stimulated cell cytokines release (e.g., release of IL-6, IL10, IL12p70, IL23, IL8 and TNF- ⁇ ) from peripheral blood monocytes, DCs and/or monocytes-derived DCs stimulated with Pansorbin or soluble CD40L and IFN′. The activity of these molecules is completely different from the activity of anti-SIRPa antibodies disclosed in the present application.
  • WO2013/056352 describes the use of an antibody anti-human SIRPa (called 29AM4-5, and corresponding to SIRP-29 as named by the inventors of the present invention) in a model of SIRPa-positive AML, i.e. xenotransplantation into immunodeficient mice of primary human AML cells expressing human SIRPa to which bind said antibody anti-human SIRPa.
  • the approach consists in the use of an anti-human SIRPa antibody to act on tumor cells expressing human SIRPa. The treatment is thus directly directed toward the tumor.
  • WO 2015/138600 describes anti-human SIRPa antibodies that bind to human SIRPa and block the interaction with CD47 expressed on a target cell with SIRPa expressed on a phagocytic cell. This document does not suggest nor show any evidence of the effect of an anti-SIRPa on macrophage polarization nor on the increase of the pro-inflammatory function of macrophages.
  • the inventors named in WO 2015/138600 published an abstract at the 56th ASH annual meeting (Weiskopf et al, ASH 2014). In this abstract, which is posterior to the filing of WO 2015/138600, they mentioned that anti-human SIRPa antibodies blocking the interaction between CD47 and SIRPa were not sufficient to induce human macrophage phagocytosis.
  • these publications suggest that the mechanism of action of an anti-human SIRPa in vivo is different from that of an anti-human CD47, for which the in vivo phagocytic efficacy was largely described in the literature.
  • the inventors provide a new insight in the use of anti-SIRPa compounds since the modulation of the immune environment is a major achievement in the treatment of many diseases, especially in cancer.
  • One advantage of the invention is that the use of an anti-SIPRa compound will induce less side effects than the use of an anti-CD47 compound.
  • CD47 is expressed by a large range of cells, and not only by tumor cells and interacts with several ligands.
  • the expression of SIRPa being more limited, the effect of the therapy will be more targeted on the tumor microenvironment.
  • the use of an anti-SIRPa compound will be less toxic and less deleterious than the use of an anti-CD47 compound.
  • the therapy being directed toward macrophages and not tumor cells, no selection pressure will be exercised on tumor cells allowing to prevent tumor escape and the development of tumor resistance to the treatment.
  • the present invention hence pertains to the use of an anti-SIRPa compound able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophage, such as an anti-SIRPa antibody, for modifying macrophage polarization.
  • the method of the invention thus consists in the use of an anti-SIRPa compound wherein said compound inhibits polarization of M2-type macrophages and/or favors pro-inflammatory M1-type of macrophages.
  • said anti-SIRPa compound can be selected from the group consisting of an anti-SIRPa antibody, in particular an anti-SIRPa antagonist antibody, a nucleic acid encoding such compound, and a compound able to inhibit the expression of the SIRPa protein, in particular a siRNA.
  • Anti-SIRPa compounds in particular anti-SIRPa specific antibodies, can thus be used in the treatments of various conditions likely to be improved or prevented by pro-inflammatory macrophages, such as cancers, infectious diseases, traumas, auto-immune diseases, vaccination, neurologic diseases, brain and nerve injuries, polycythemias, hemochromatosis and chronic inflammatory diseases. Cancer is a preferred therapeutical indication.
  • pro-inflammatory macrophages such as cancers, infectious diseases, traumas, auto-immune diseases, vaccination, neurologic diseases, brain and nerve injuries, polycythemias, hemochromatosis and chronic inflammatory diseases.
  • cancer is a preferred therapeutical indication.
  • the present invention concerns an anti-SIRPa compound able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophage for use in the treatment of cancer, with the exception of SIRPa-positive acute myeloid leukemia and/or SIRPa-positive non acute myeloid leukemia and/or SIRPa-positive non-Hodgkin leukemia or SIRPa-positive hematologic cancers.
  • the therapeutical approach aims to target SIRPa on the macrophage in order to modulate their polarization and to recreate an immune context detrimental to the tumor development and survival.
  • the success on the anti-tumor treatment is based on an indirect pathway, and does not require that the tumor cells are sensitive to the anti-SIRPa compound.
  • the present invention concerns the use of an anti-SIRPa compound as defined herein, i.e. able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophage, in the treatment of cancer, wherein said compound is administered to a patient presenting a SIRPa-negative tumor.
  • Anti-SIRPa compounds as defined herein can be used in monotherapy as described in the experimental part.
  • Combinations of anti-SIRPa compounds as defined herein with other therapeutic agents, especially with agents blocking another immune checkpoint, are also part of the invention, since a synergistic effect was demonstrated by the inventors.
  • Another aspect of the present invention is a method for ex vivo obtaining pro-inflammatory M1-type macrophages by incubating macrophages with an anti-SIRPa compound as defined herein.
  • the present invention also pertains to a method for selecting patients likely to benefit from a treatment by an anti-SIRPa compound as defined herein, by measuring the presence of M2-type macrophages in a sample from the patient.
  • a method of following-up a treatment by an anti-SIRPa compound as defined herein, to assess its efficacy by measuring the presence of pro-inflammatory M1-type macrophages and/or measuring the presence of anti-inflammatory M2-type macrophages in a sample from an individual treated by said compound, is also part of the present invention.
  • FIG. 1 Monocyte type 1 polarization with GM-CSF+M-CSF protocol: Effect of SIRPa blockade: Human Monocytes were cultivated with growth factors M-CSF and GM-CSF to induce Macrophages and treated or not with Ctrl Ab or anti-Sirp antibodies (anti-a or ab or b isotype), or a CD47-Fc protein, or some anti-CD47 antibodies (B6H12 or CC2C6). Then type 1 macrophage phenotype and function were analysed by FACS and ELISA. A. represents cell surface markers analysis: CD86 and CCR7. B. represents secreted cytokine/chemokine: IL6, IL12p40, CCL-2 and TNF- ⁇ . IL6, p12p40 and CCL-2 are significantly increased when cells are treated with an anti-SIRPa antibody.
  • SIRPa blockade Human Monocytes were cultivated with growth factors M-CSF and GM-CSF to induce Macrophages and
  • FIG. 2 Monocyte Type 2 polarization with GM-CSF+M-CSF protocol: Effect of SIRPa blockade: Human Monocytes were cultivated with growth factors M-CSF and GM-CSF to induce macrophages and treated or not with Ctrl Ab or anti-Sirp antibodies (anti-a or ab or b isotype), or a CD47-Fc protein, or some anti-CD47 antibodies (B6H12 or CC2C6). Then Type 2 macrophage phenotype and functional analysis were performed by FACS and ELISA. A. Shows marker expression at cell surface: CD206, CD200R, CD11b and B. Shows secreted Chemokine: CCL-17
  • FIG. 3 Monocyte type 1 polarization with M-CSF and IFNgamma protocol: Effect of SIRPa blockade: Human monocytes were cultivated 5-6 days with M-CSF and then 2 days with IFNgamma with or without Ctrl Ab or anti-Sirp antibodies (anti-a or ab or b Sirp isotype), or a CD47-Fc protein, or some anti-CD47 antibodies (B6H12 or CC2C6). M1 secreted cytokines were analysed: IL6 and IL12p40. Statistical analysis were performed
  • FIG. 4 Monocyte type 1 polarization with M-CSG+IFNgamma and LPS protocol: Effect of SIRPa blockade on iNOS expression: Human monocytes were cultivated 5-6 days with M-CSF and then 2 days with IFNgamma+LPS with or without antibodies directed against: SIRPa: SE7C2 (A.) or SIRPa/b: SE5A5 (B.) or CD47: B6H12 (C.), iNOS expression was then analysed by FACS. Ctrl Ab is represented by dotted line and monocytes before polarization by empty grey line on the three panels
  • FIG. 5 Monocyte type 2 polarization with M-CSF+IL4 protocol: Effect of SIRPa blockade with antibody: Human monocytes were cultivated 5-6 days with M-CSF and then with IL4 with or without Ctrl Ab or anti-Sirp antibodies (anti-sirp a or ab or b), or a CD47-Fc fusion protein, or some anti-CD47 antibodies (B6H12 or CC2C6).
  • A. Represents expression of cell surface markers: CD206, CD200R and CD11b and B. Represents the release of IL6 cytokine in the supernatant
  • FIG. 6 Monocyte type 2 polarization with M-CSF+IL4 protocol: Effect of SIRPa blockade at a mRNA level with a siRNA Knock down assay: Human monocytes were cultivated 5-6 days with M-CSF and then 2 days with IL4. Cells were transfected with null-siRNA or SIRPa siRNA: A. represents the expression of the cell surface markers: CD206 and CD200R; B. represents the IL6 cytokine release in the supernatant.
  • the anti-SIRPa SiRNA are provided by ThermoFisher scientific.
  • FIG. 7 Effect of SIRPa blockade on M2 repolarization after M-CSF+IL4 treatment: Human monocyte were cultivated 5-6 days with M-CSF and then 2 days with IL4. M2 macrophages were then treated with or without Ctrl Ab or anti-Sirp antibodies (anti-sirp a or ab or b), or a CD47-Fc fusion protein, or some anti-CD47 antibodies (B6H12 or CC2C6). The Cytokines IL6 and TNF- ⁇ releases were then measured in the supernatant.
  • Anti-SIRPa+anti-CD137 grey diamond; n
  • FIG. 10 Effect of a combined anti-SIRPa+anti-PD-L1 treatment on an in vivo model of melanoma:
  • FIG. 11 Effect of anti-SIRPA antibody on mouse macrophage polarization: Mouse M2 macrophage polarization phenotype is inhibited by anti-SIRPa mAb but not by anti-CD47 mAb as shown by the quantification of the expression of the M2 markers CD206, CD11b and PD-L1.
  • anti-SIRPa mAb corresponds to p84 clone and anti-CD47 mAb corresponds to MIAP310 clone.
  • FIG. 12 Effect of anti-SIPRa antibody on human M2 macrophage polarization: Human M2 macrophage polarization phenotype is inhibited by anti-SIRPa mAbs but not with anti-CD47 mAb.
  • A. By detection of M2 surface markers (CD200R, CD80).
  • B. At functional level by evaluation of cytokine secretion (IL-6).
  • IL-6 cytokine secretion
  • FIG. 13 Effect of anti-SIPRa antibody on human M1 phagocytosis: Human macrophage phagocytosis of CD47+tumor cells (Raji) is increased by CD47 targeting agents, but not with SIRPa targeting agents. Analysis of phagocytosis was evaluated by flow cytometry by gating on M1 fluorescent macrophages and analysis of target (Raji) cells fluorescence into M1 macrophages.
  • the CD47 targeting agents correspond to anti-CD47 mAbs B6H12 or CC2C6 clones and the SIRPa targeting agents correspond to anti-SIRPa clone SE7C2 or CD47-Fc protein.
  • FIG. 14 Effect of anti-SIRPA antibody on rat M1/M2 polarization: A. Rat M2 macrophages become pro-inflammatory and secrete inflammatory cytokines (IL-6 and TNF- ⁇ ) in presence of anti-SIRPa mAb, but not anti-CD47 mAb. B. M1 rat macrophages polarization is not modified by anti-SIRPa or anti-CD47 mAbs. Anti-SIRPa mAb used in this experiment corresponds to ED9 clone and anti-CD47 mAb corresponds to OX101 clone.
  • FIG. 15 Effect of anti-SIRPa antibody on a mouse model of mammary tumor.
  • Monotherapy with anti-SIRPa mAb inhibit tumor growth in the 4T1 mammary cancer model in mice.
  • Anti-SIPRa mAb used in this experiment corresponds to clone p84.
  • the term “polarization” is used herein to designate the phenotypic features and the functional features of the macrophages.
  • the phenotype can be defined through the surface markers expressed by the macrophages.
  • the functionality can be defined for example based on the nature and the quantity of chemokines and/or cytokines expressed, in particular secreted, by the macrophages. Indeed, the macrophages present different phenotypic and functional features depending of their state, either pro-inflammatory M1-type macrophage or anti-inflammatory M2-type macrophage.
  • M2-type macrophages can be characterized by the expression of surface markers such as CD206, CD11b, PD-L1 and CD200R and then secretion of cytokines such as CCL17.
  • M1-type macrophages can be defined by the expression of surface markers such as CD86 and CCR7 and the secretion of cytokines such as IL-6, TNF- ⁇ and IL12p40.
  • anti-SIPRa compounds allow to modulate the polarization of macrophages population by inhibiting the M2-type macrophages and/or favoring the M1-type macrophages.
  • activation usually used to mean the perturbation of macrophages with exogenous agents. Macrophages change their polarization states in response to growth-factors (CSF-1 and GM-CSF) and external stimuli such as microbes, microbial product and nucleotides derivatives, antibody-Fc receptor stimulation, glucocorticoids, phagocytosis.
  • CSF-1 and GM-CSF growth-factors
  • GM-CSF growth-factors
  • external stimuli such as microbes, microbial product and nucleotides derivatives, antibody-Fc receptor stimulation, glucocorticoids, phagocytosis.
  • an “anti-SIRPa compound”, as used herein refers to a compound that specifically binds to the extracellular domain of SIRPa, or a nucleic acid encoding such compound, as well as to a compound able to inhibit the expression of the SIRPa protein. Such compound is able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophage.
  • An anti-SIRPa compound can be a compound specifically binding to the extracellular domain of the signal regulatory protein alpha (SIRPa).
  • An anti-SIRPa compound can also be a SIRPa-blocking compound.
  • an anti-SIRPa compound is an antagonist peptide or an anti-SIRPa antibody, in particular an anti-SIRPa antagonist antibody.
  • an antibody refers to polyclonal antibody, monoclonal antibody or recombinant antibody.
  • the monoclonal antibodies of the present invention include recombinant antibodies for example, chimeric, CDR graft and humanized antibodies, but also antigen-binding moiety.
  • the term “antigen-binding moiety” of an antibody refers to one or more fragments of an antibody of the invention, said fragment(s) still having the binding affinities as defined above.
  • examples of binding fragments include Fab fragment, F(ab′)2 fragment, Fv fragment, and single chain Fv (ScFv). Other forms of single chain antibodies such as “diabodies” are likewise included here.
  • an antagonist peptide refers to a peptide able to inhibit the interaction of SIRPa with one of its ligands, especially with CD47, or to a peptide able to prevent or decrease the SIRPa signaling pathway.
  • An anti-SIRPa compound can also be a compound able to inhibit the expression of the SIRPa protein such as an antisens oligonucleotide, or an interfering RNA such as a shRNA or a siRNA.
  • siRNA able to modulate the polarization of the macrophages according to the invention are provided in the experimental part. Further, a skilled person in the art knows how to identify such compound.
  • an anti-SIRPa compound able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophage can be identified by applying one of the protocols described in the experimental part below.
  • an anti-SIRPa X compound can be performed as follows:
  • M0 macrophages are obtained by culturing monocytes 5 to 6 days with M-CSF (100 ⁇ g/mL), then cells are cultured in vitro during 2 days with recombinant hIL4 (20 ng/mL) in the presence or in the absence of said compound X.
  • anti-SIRPa mAb with a known activity can be used. Any compound not targeting the extracellular domain of SIRPa can be used as a negative control (e.g., an anti-SIRPb antibody or a CD47-Fc).
  • the secretion of different cytokines, for example IL-6, TNF- ⁇ and IL12 and chemokine CCL17 can be measured by ELISA.
  • the compound X can be classified as an “anti-SIRPa compound” if (i) it inhibits hallmarks of M2 macrophage features, in particular the overexpression of surface markers such as CD206, CD11b, PD-L1 and/or CD200R, and the secretion of specific cytokines such as CCL17 and/or (ii) it increases hallmarks of M1 macrophage features, in particular the expression of surface markers such as CD86 and CCR7 and the secretion of cytokines such as IL-6, TNF- ⁇ , IL12-p40 and/or IL12, as efficiently or more efficiently than the positive control mAb.
  • the effect of an anti-SIRPa compound can be evaluated in comparison with the effect of the particular antibody SE7C2 (Santa Cruz sc-23863), used at a concentration of 10 ⁇ g/mL.
  • SE7C2 Santa Cruz sc-23863
  • the compound X is classified as an “anti-SIRPa compound” if it the results of the testing show that it is as efficient as SE7C2, or more efficient than SE7C2.
  • the comparison can be done with any anti-SIRPa compound of reference, as a positive control.
  • cancer means all types of cancers.
  • the cancers can be solid or non-solid cancers.
  • Non limitative examples of cancers are carcinomas or adenocarcinomas such as breast, prostate, ovary, liver, lung, bladder, pancreas or colon cancer, sarcomas, lymphomas, melanomas, leukemias, germ cell cancers and blastomas.
  • the terms “treat”, “treatment” and “treating” refer to any reduction or amelioration of the progression, severity, and/or duration of cancer, particularly a solid tumor; for example in a liver cancer, reduction of one or more symptoms thereof that results from the administration of one or more therapies.
  • a “therapeutic agent” designates any active force or substance capable of producing an effect. Therapeutic agents thus include radiations, surgery, probiotics as well as any kind of drug.
  • a “drug blocking an immune checkpoint”, or “immune checkpoint blocker” or “immune checkpoint blockade drug” or “immune checkpoint inhibitor” designates any drug, molecule or composition which blocks an immune checkpoint.
  • it encompasses an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody and an anti-SIRPa antibody.
  • two or more immune checkpoint inhibitors can be combined to target both adaptive or innate immune cells. This approach is particularly interesting in the treatment of cancer.
  • an anti-SIRPa compound can be combined with an anti-PD-L1 compound. Such combinations can be used simultaneously separately or sequentially, particularly, in the treatment of cancer.
  • immune checkpoint stimulator designates any drug, molecule or composition which activates an immune checkpoint.
  • Such molecules or drugs that stimulate costimulatory molecules, resulting in target cell activation are for example anti-CD137 antibodies.
  • a first aspect of the present invention is the use of an anti-SIRPa compound able to modify macrophage polarization, in particular able to inhibit the polarization of anti-inflammatory M2-type macrophages and/or favors pro-inflammatory M1-type macrophage, in an individual (e.g., a human) in need thereof.
  • macrophages mature in tissues and are activated in a dynamic response to combinations of stimuli to acquire specialized functional phenotypes that, in certain cases, are detrimental to the individual.
  • M1 vs. alternative (M2) a dichotomy has been proposed for macrophage activation: classic (M1) vs. alternative (M2).
  • M1 and M2 subtypes remain relevant to describe the extremes on a continuum of macrophage states, M1 being the most pro-inflammatory state and M2 being more associated with a decrease of inflammation.
  • modifying macrophage polarization it is herein meant that the compound modifies the balance between the different subtypes of macrophages in the treated individual, at least at the phenotypic and/or functional level.
  • the treatment of an individual with an anti-SIRPa compound leads to a modification in the profile of surface markers expressed by the individual's macrophage (including a decrease of CD206, CD11b, PD-L1 and/or CD200R expression and an increase of CD86 and CCR7 expression) and the profile of chemokines and cytokines expressed and/or secreted by the individual's macrophages (including a decrease of CCL-17 expression and an increase of IL6, TNF- ⁇ and IL12p40 expression).
  • the modification of macrophage polarization by the anti-SIRPa compound includes an inhibition of M2-type macrophage polarization and/or an increase of pro-inflammatory M1-type macrophage polarization.
  • it can include an inhibition of M2 phenotypic polarization of macrophages, leading to a decrease of the proportion of macrophages overexpressing cellular markers such as CD206, CD11b, PD-L1 and/or CD200R and/or producing cytokines such as CCL17.
  • the anti-SIRPa compound can induce the emergence of more macrophages exhibiting a M1 phenotypic polarization and an increase of the proportion of macrophages overexpressing cellular markers such as CD86 and CCR7 and/or producing cytokines such as IL-6, IL-12p40 and/or TNF- ⁇ .
  • the anti-SIRPa compound can thus modulate the macrophage polarization at both phenotypic level (expression of cellular surface markers) and functional level (production of chemokines and cytokines).
  • the compound used for modifying macrophage polarization is an anti-SIRPa compound.
  • An exemplary test to determine if a given compound is an anti-SIRPa compound in the sense of the present application is described above.
  • anti-SIRP antibodies such as those used in the experiments described below, any other antibody selected amongst the many anti-SIRPa commercially available antibodies or any other (new) anti-SIRPa antagonist antibody, fragments of antibodies, aptamers targeting SIRPa, etc.
  • the present invention also pertains to the use of a nucleic acid (mRNA or DNA) encoding an anti-SIRPa compound, for modifying macrophage polarization to favor M1 pro-inflammatory macrophages in an individual in need thereof.
  • a nucleic acid mRNA or DNA
  • administering a nucleic acid which leads to the expression of an anti-SIRPa compound, as above-described by the patient's cells is an alternative possibility for obtaining the desired effect on macrophage polarization.
  • anti-SIRPa compound is herein considered to encompass nucleic acids enabling the in vivo expression of such a compound.
  • the anti-SIRPa compound is selected from the group consisting of an antagonist peptide, an anti-SIRPa antibody, in particular an anti-SIRPa antagonist antibody, a nucleic acid encoding such compound, and a compound able to inhibit the expression of the SIRPa protein, in particular a siRNA.
  • an anti-SIRPa compound is an antagonist peptide or an anti-SIRPa antibody, in particular an anti-SIRPa antagonist antibody.
  • Modifying the polarization of macrophages to favor M1 pro-inflammatory cells can be useful in a number of pathologies or situations. As described above, this modification is particularly useful in the context of cancers, to restore an anti-tumor activity of macrophages and/or prevent the pro-tumoral activity of M2-type macrophages. Indeed, immune responses due to an excess of M2-type macrophage polarization also occur in infectious diseases, vaccination, trauma and chronic inflammatory diseases.
  • Macrophages are supposed to interact with stem cells or progenitor cells to control repair and remodeling functions.
  • Cells from macrophages lineage present some neuroprotective effect.
  • Mesenchymal cells (MSC) are targeted to promote tissue repair and immunoregulation. Injection of MSC was associated with a benefit for the recovery of functions of the spinal cord injury such as axonal preservation and reduced scare formation.
  • the neuroprotective effect was attributed to a polarization shift from M2 to M1 macrophages by MSC (Nakajima et al., 2012), indicating that any molecules enabling this shift, such as an anti-SIRPa compound, could have a neuroprotective effect.
  • Macrophages are also involved in some iron deficiencies such as hemochromatosis, where the iron homeostasis is clearly impaired.
  • Polycythemia vera or essential polycythemia rubra is a myeloproliferative disorder characterized by polycythemia (significant increase in the number of red blood cells) and an increase in the total cell volume. Red cells subsequently pass into the blood and could progress into a myeloproliferative syndrome.
  • Patient treated with an anti-CD47 present anemia indicating that the blockade of CD47 is not as safe as expected; targeting SIRPa through an anti-SIRPa compound in the sense of the present invention could avoid this side effect.
  • the present invention thus pertains to the use of an anti-SIRPa compound, for modifying macrophage polarization to favor M1 pro-inflammatory macrophages in an individual suffering from a cancer, an infectious disease, a trauma, an auto-immune disease, a neurologic disease, a brain injury, a nerve injury, a polycythemia, a hemochromatosis or a chronic inflammatory disease, as well as in a context of vaccination of an individual.
  • the anti-SIRPa compound is used to treat an individual who has a cancer selected from the group consisting of lung cancers, ovary cancers, liver cancers, bladder cancers, brain cancers, breast cancers, colon cancers, thymomas, gliomas, melanomas and hematologic cancers.
  • the anti-SIRPa compound is used in the treatment of any cancer with the exception of SIRPa-positive acute myeloid leukemia (AML).
  • AML acute myeloid leukemia
  • the treatment of tumor cells involved in AML, which express SIRPa is thus directed toward the tumor cells, i.e. through a direct targeting of the tumor.
  • This therapeutical strategy is thus different from the indirect approach proposed in the present invention.
  • the anti-SIRPa compound is used in the treatment of any cancer with the exception of SIRPa-positive acute myeloid leukemia and SIRPa-positive non acute myeloid leukemia.
  • the anti-SIRPa compound is used in the treatment of any cancer with the exception of SIRPa-positive non-Hodgkin lymphoma or non-Hodgkin lymphoma. In a further particular embodiment, the anti-SIRPa compound is used in the treatment of any cancer with the exception of SIRPa-positive acute myeloid leukemia and/or SIRPa-positive non acute myeloid leukemia and/or non-Hodgkin lymphoma or hematologic cancers.
  • the anti-SIRPa compound is used in the treatment of any cancer with the exception of i) SIRPa-positive acute myeloid leukemia or acute myeloid leukemia, and/or ii) SIRPa-positive non acute myeloid leukemia or non acute myeloid leukemia, and/or iii) SIRPa-positive non-Hodgkin lymphoma or non-Hodgkin lymphoma, or iv) SIRP-a positive hematologic cancers or hematologic cancers.
  • the anti-SIRPa compound is used in the treatment of cancer with SIRPa-negative tumor cells, as described thereafter in the description.
  • the anti-SIRPa compound is used in a monotherapy, in particular in the treatment of breast cancer, hepatocarcinoma or melanoma.
  • Modulation macrophage polarization is a very attractive approach to treat cancer especially in a combined therapy of cancer.
  • Antonia et al. (Antonia et al., 2014) defined immuno-oncology combination that could be interesting for cancer treatment using checkpoint inhibitor approaches.
  • Immuno-oncology is an evolving treatment modality that includes immunotherapies designed to harness the patient's own immune system.
  • an anti-SIRPa compound in the sense of the present invention, can be combined with some other potential strategies for overcoming tumor immune evasion mechanisms with agents in clinical development or already on the market (see table 1 from (Antonia et al., 2014)):
  • an anti-SIRPa compound can also be combined with a microbiome-modulating strategy to improve the anti-cancer immune response.
  • Another aspect of the present invention is thus the use of an anti-SIRPa compound as above-defined, in combination with a second therapeutic agent, to treat an individual in need thereof, in particular a cancer patient.
  • a second therapeutic agent to treat an individual in need thereof, in particular a cancer patient.
  • Such combinations can be used simultaneously separately or sequentially, particularly, in the treatment of cancer.
  • the second therapeutic agent is selected from the group consisting of chemotherapeutic agents, radiotherapy, surgery, immunotherapeutic agents, antibiotics and probiotics.
  • the second therapeutic agent can advantageously be selected from the group consisting of therapeutic vaccines, immune checkpoint blockers such as, for example, anti-PDLL, anti-PD1 and anti-CTLA4 and immune checkpoint activators such as, for example, anti-CD137.
  • immune checkpoint blockers such as, for example, anti-PDLL, anti-PD1 and anti-CTLA4
  • immune checkpoint activators such as, for example, anti-CD137.
  • these combinations produce synergistic effects.
  • on aspect of the invention consists in the use of an anti-SIRPa compound in combination with a second immune checkpoint modulator in the treatment of cancer selected among hepatocarcinoma or melanoma.
  • the anti-SIRPa mAb is combined with an anti-CD137 mAb in the treatment of hepatocarcinoma.
  • the anti-SIRPa mAb is combined with an anti-PD-L1 mAb in the treatment of melanoma.
  • Another aspect of the present invention is a method for ex vivo obtaining pro-inflammatory M1-type macrophages, comprising a step of incubating macrophages with an anti-SIRPa compound as described above.
  • This method can be useful, for example, in cell therapy, especially for cancer patients.
  • the present text also describes a method for treating a cancer patient, comprising a step of obtaining macrophages from said patient, followed by a step of modifying their polarization to favor M1-associated pro-inflammatory functions through incubation with an anti-SIRPa compound, and a step of re-administering the obtained pro-inflammatory cells to the patient.
  • additional steps such as expanding the cells, selecting the cells which exhibit a M1-type phenotype and/or counter-selecting those which exhibit a M2-type phenotype
  • additional steps can be introduced in such a method.
  • the present invention pertains to a method for in vivo determining the efficacy of a treatment by an anti-SIRPa compound as defined above, comprising measuring the presence of pro-inflammatory M1-type macrophages and/or measuring the presence of M2-type macrophages in a sample from an individual treated by said compound.
  • the presence of pro-inflammatory macrophages can be measured, for example, by measuring the levels of IL6, TNF- ⁇ and/or IL12p40 secreted by the macrophages present in the sample.
  • the presence of M2-type macrophages can be measured, for example, by measuring the expression of CD206, CD11b, PD-L1 and/or CD200R on the surface of macrophages.
  • This therapeutic approach could not have been envisaged before, as prior art taught that the treatment of cancer relied on the inhibition of the SIRPa-CD47 interaction at tumor cell level (especially by inducing phagocytosis).
  • the inventors of the present invention demonstrated that, on the contrary, anti-SIRPa mAb do not induce phagocytosis and act on a distinct pathway.
  • anti-SIPRa compounds target the M2-type macrophages and allow to repolarize this population of macrophages into M1-type macrophages.
  • the proposed invention responds to a “new clinical situation” not described before.
  • the anti-SIRPa compound of the invention allows to treat cancer by indirect approach, targeting the innate immune system, in particular by inhibiting the inhibition of inflammation in tumor environment, and more specifically by inhibiting the anti-inflammatory M2-type macrophages.
  • an object of the invention consists in the use of an anti-SIRPa compound in the treatment of cancer, wherein the said compound is administered to a patient presenting a SIRPa-negative tumor.
  • a “SIRPa-negative tumor” corresponds to a tumor containing a SIRPa-negative cellular population.
  • this term encompasses both a tumor consisting in SIRPa-negative cells and a tumor which may contain a mixed population of SIRPa-positive tumor cells and SIRPa-negative tumor cells.
  • the present invention also concerns the use of said compound for treating cancer wherein the tumor of the patient comprises a mixed population of tumor cells containing both SIRPa-positive and SIRPa-negative cells.
  • the present invention also provides a method for treating cancer comprising the administration of an anti-SIRPa compound able to inhibit the polarization of anti-inflammatory M2-type macrophage and/or to favor pro-inflammatory M1-type macrophage, to a patient in need thereof.
  • the present invention also concerns a combination product comprising:
  • the half product is used to treat cancer, with the exception of SIRPa-positive acute myeloid leukemia.
  • the present invention hence also pertains to a method for in vivo determining if an individual is likely to be a good responder to a treatment by an anti-SIRPa compound in the sense of the present invention, comprising measuring the presence of M2-type macrophages in a sample from said individual, for example by measuring the expression of CD206, CD11b, PD-L1 and/or CD200R on the surface of macrophages present in the sample.
  • the sample used either to assess whether an individual is likely to be a good responder to a treatment with an anti-SIRPa compound or to determine the efficacy of such a treatment can be a blood sample, a tissue sample, a sample from a tumor or a sample of synovial liquid.
  • PBMC peripheral blood Mononuclear cells
  • SIRPa mAbs SE7C2 (Santa Cruz sc-23863) or clone p84 (Merck Millipore); Sirp ⁇ / ⁇ mAbs: SE5A5 (BioLegend BLE323802); anti-CD47: B6H12 (eBioscience 14-0479-82) or CC2C6 (BioLegend BLE323102); CD47-Fc: SinoBiological 12283-H02H; anti-CD137 antibody (clone 3H3 produced in-house) and anti-PD-L1 antibody (10F-9G2 from BioXCell)
  • M1/M2 differentiation in the same well was realised by culturing monocytes in 24-well plates at 4.10 5 cells/well in complete RPMI with GM-CSF (10 ng/mL—CellGenix) for 3 days and then with GM-CSF and M-CSF (2 ng/ml and 10 ng/mL, respectively—R&D systems) for 3 supplementary days (Haegel et al., 2013).
  • Antibodies were added at day 0 and day 3. At day 6, cells were harvested and were analysed by Flow Cytometry using antibodies from BD Bioscience.
  • In vitro mouse macrophages differentiation was performed by culturing cells from the bone marrow in 24-well plates at 0.5.10 6 cells/mL in complete RPMI for 4 days supplemented by murine M-CSF (100 ⁇ g/mL—Peprotech) to induce M0 macrophages.
  • M2 polarization is induced by murine IL-4 (20 ng/mL—Peprotech) to generate M2 anti-inflammatory macrophages for 24 hours.
  • Analysis of phenotype changes was performed by flow cytometry staining using antibodies from BD Bioscience.
  • iNOS Measurement by Multiplexed Nanoflares iNOS expression was revealed by the SmartFlare technology (Prigodich et al., 2012). Briefly, monocytes or macrophages were collected and incubated with the iNOS probe (1 nM final) 2 h at 4° C., washed and analysed on a LSR II (BD).
  • siRNA coding for endogenous SIRPa were transfected into macrophages (M0-macrophages pre-polarized by M-CSF). Three sequences of siRNA (ThermoFisher Scientific, ref:112328, 112327 and 109944) were chosen and pooled to downregulate SIRPa expression. In a 24-well plate, 90 pmol of siRNA-SIRPa (3*30 pmol of each siRNA-SIRPa) were diluted in 100 ⁇ l Opti-MEM Medium without serum and mixed gently. Then 1 ⁇ l/well of lipofectamine RNAiMAX (ThermoFisher Scientific, ref 13778-150) was mixed and incubated for 20 min. at room temperature.
  • M2 were plated at 100 000 cells/ml in 500 ⁇ l of complete growth medium with IL4 (20 ng/ml)+/ ⁇ 100 ⁇ l of the siRNA-lipofectamine complexes. Null-siRNA was used as a control of transfection. Cells were incubated 48 hours at 37° C., 5% CO 2 .
  • Cytokines and chemokines released in the supernatant were analyzed by Elisa using materials of BD Bioscience and R&D systems (references below), according to the manufacturer's instructions. Supernatants were diluted at 1/200.
  • PK136 anti-NK1.1 (mouse mAb IgG2a) and SF1-1.1 anti-H2Kd (mouse mAb IgG1) were used as isotypic control antibodies.
  • Human M1 pro-inflammatory macrophages were generated as previously described and stained with a fluorescent dye.
  • the CD47-expressing Raji cells were stained with another fluorescent dye and incubated with M1 stained macrophages during 2 h at 37° C.
  • Cells were fixed with paraformaldehyde and analysis of phagocytosis was evaluated by flow cytometry by gating on M1 fluorescent macrophages and analysis of target (Raji) cells fluorescence into M1 macrophages.
  • mice received 2.5 ⁇ 10 6 Hepa1.6 mouse hepatoma cells in 100 ⁇ L through the portal vein, as previously described (Gauttier et al., 2014).
  • mice were injected intraperitoneally with 100 ⁇ g of rat anti-4-1BB mAb (clone 3H3 produced in house), or with 300 ⁇ g of anti-mouse SIRPa monoclonal antibody (clone P84 from Merck Millipore) or both antibodies or an irrelevant control antibody (clone 3G8) 3 times/week for 4 weeks or with 200 ⁇ g of the anti-PD-L1 mAb (clone 10F-9G2 from BioXCell) or received both antibodies (anti-Sirpa+anti-PDL1) for 4 weeks.
  • mice received subcutaneous injection of 2 ⁇ 10 6 B16-Ova mouse melanoma cells into the flank. Mice were treated i.p. from day 0 after tumor inoculation with either 300 ⁇ g of an irrelevant control antibody (clone 3G8) or anti-mouse SIRPa monoclonal antibody (clone P84) 3 times per week or with 200 ⁇ g of the anti-PD-L1 mAb (clone 10F-9G2 from BioXCell) twice a week or received both antibodies (anti-Sirpa and anti-PD-L1 antibodies) for 4 weeks. Some animals were sacrificed at two weeks after tumor inoculation to characterized tumor leukocytes infiltrates by flow cytometry. The overall survival was analyzed.
  • mice Eight-weeks-old Balb/c fenate mice were injected with 0.25.106 4T1 (mammary carcinoma) cells in the mammary gland in 50 ⁇ L. Mice were treated i.p. from day 4 after tumor injection with either 300 ⁇ g of an irrelevant control antibody (clone 3G8) or the anti-mouse SIRPa blocking antibody (clone P84) three times a week and during four weeks. Mice were euthanized six weeks after tumor inoculation. Tumor measurement was performed every 2-3 days and the tumor volume was determined according the calcul: length*width*Pi/6 (mm3).
  • FIG. 1A shows that antibodies directed against SIRP molecules or CD47 do not prevent M1 macrophage polarization induced by GM-CSF+M-CSF because the expressions of the M1 cell surface markers (CD86 and CCR7) are not modified compared to control conditions.
  • the over-expression of CD206, CD200R, CD11b and PD-L1 was significantly inhibited with selective anti-SIRP alpha mAb ( FIG. 2A and FIG. 11 ) but not with control antibodies (anti-SIRPa/b or Sirpb), CD47-Fc recombinant protein or anti-CD47 mAbs (clones B6H12 and CC2C6).
  • FIG. 1A shows that antibodies directed against SIRP molecules or CD47 do not prevent M1 macrophage polarization induced by GM-CSF+M-CSF because the expressions of the M1 cell surface markers (CD86 and CCR7) are not modified compared to control conditions.
  • cytokines and chemokines secretion showed that anti-SIRPa mAb prevented CCL-17 secretion (a hallmark chemokine of M2 secretion) while they increased secretion of pro-inflammatory cytokines (IL-6, IL12p40, TNF- ⁇ ) and chemokine (CCL-2) secreted by M1 macrophages ( FIGS. 1B and 2B ).
  • Anti-SIRPa thus seems to play a role on the prevention of M2 polarization and on the pro-inflammatory function of the macrophages.
  • SIRPa The role of SIRPa on M2 polarization was then studied at a transcriptional level with SIRPa inhibition, using a cocktail of siRNAs.
  • the results, presented in FIG. 6A confirmed that inhibiting SIRPa inhibits M2 polarization of the cells (CD206 and CD200R expression).
  • Maximal/terminal M1 polarization is considered achieved using M-CSF+IFNg+LPS, which results in all hallmarks of M1 macrophage, in particular iNOS expression.
  • FIG. 4 shows the expression of iNOS in M1 polarized cells treated or not with different blocking antibodies.
  • blocking SIRPa with an antibody increased the expression of iNOS.
  • anti-SIRP13 or anti-CD47 mAb did not induce any modification in the iNOS expression profile.
  • the phenotype and function of polarized M1/M2 macrophages has been, to some extent, reversed in vitro or in vivo due to the plasticity of these cells (Sica and Mantovani, 2012).
  • the inventors addressed the question of the repolarization of the M2 type into a pro-inflammatory M1 type by blocking SIRPa. To do so, monocytes were cultivated with M-CSF+IL-4 inducing M2 terminal polarization. Then M2 cells were treated with different blocking antibodies. Results presented FIG.
  • results of FIG. 12 show that two different anti-SIRPa mAb allow to induce the polarization of macrophage in favor of M2-type macrophages, both at phenotypic level with expression of surface markers CD200R and CD80 (A) and at functional level with secretion of IL-6 (B).
  • results of FIG. 14 show that the selective blockade of SIRPa but not CD47 prevents M2 polarization (B) and did not affect M1 polarization (A).
  • B M2 polarization
  • A M1 polarization
  • the specific blockade of SIRPa (but not with anti-CD47) during M2 polarization of rat macrophages switches their cytokine profile towards an inflammatory profile (like M1 macrophages) with the secretion of IL-6 and TNF- ⁇ .
  • SIRPa is important for the M2 polarization of the monocyte and blocking this pathway is a good opportunity to produce macrophages with pro-inflammatory profile to treat pathologies in need thereof such as cancer or for infectious diseases, in which macrophages are blocked in an M2 state.
  • FIG. 13 shows that two different anti-CD47 mAbs increase the phagocytosis of CD47+tumor cells (Raji) by M1 macrophages as described in literature.
  • the selective blockade of SIRPa by the anti-SIRPa mAb or a recombinant CD47-Fc fusion protein do not affect increase phagocytic activity of M1 macrophages.
  • FIG. 8 represents the overall survival rate of animals inoculated with hepatocarcinoma and treated with an anti-CD137, an anti-SIRPa or both during 4 weeks. 20% of the animals treated with anti-Sirpa monotherapy survived more than 25 days and 25% of animals treated with anti-CD137 monotherapy survived more than 30 days. However, 100% of the animals receiving the combo anti-Sirp+anti-CD137 were still alive after 80 days, compared to the other conditions, showing a synergistic effect of the 2 molecules.
  • FIG. 9 represents the overall survival rate of animals inoculated with hepatocarcinoma and treated with an anti-PD-L1, an anti-SIRPa or both during 4 weeks.
  • the results showed a very interesting surviving rate when animals were treated with both molecules, compared to each molecule alone (20% of alived animals after 20 days with anti-sirpa compare to 12% of alived animals with anti-PD-L1). This result indicates a synergistic effect of the anti-SIRPa antibody with the anti-PD-L1 antibody in a cancer model.
  • FIG. 10 A represents the overall survival rate of animals inoculated with melanoma cells and treated with an anti-PD-L1, an anti-SIRPa or both during 4 weeks. Compared to the treatment with each molecule alone, the combination showed a better efficacy.
  • FIG. 10 B shows the macrophage infiltrate in animals treated with anti-SIRPa, confirming an increase in macrophage number into the tumor.
  • SIRPa is an interesting target for cancer treatment, especially when combined with other immunotherapies, and suggest that Sirp is a new checkpoint inhibitor that is important to block in the aim to restore a pro-inflammatory tumor environment.
  • FIG. 15 shows that monotherapy with anti-SIRPa mAb (clone P84) inhibit tumor development in a syngeneic and orthotopic triple-negative breast model in mice. From 2 weeks post-tumor inoculation until the end of the experiment, anti-SIRPa treated mice have a significant reduction in tumor volume.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US15/769,689 2015-10-21 2016-10-21 Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer Abandoned US20180312600A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP15190918 2015-10-21
EP15190918.1 2015-10-21
PCT/EP2016/075466 WO2017068164A1 (en) 2015-10-21 2016-10-21 Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/075466 A-371-Of-International WO2017068164A1 (en) 2015-10-21 2016-10-21 Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/747,798 Continuation US20220298259A1 (en) 2015-10-21 2022-05-18 Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer

Publications (1)

Publication Number Publication Date
US20180312600A1 true US20180312600A1 (en) 2018-11-01

Family

ID=54540804

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/769,689 Abandoned US20180312600A1 (en) 2015-10-21 2016-10-21 Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer
US17/747,798 Pending US20220298259A1 (en) 2015-10-21 2022-05-18 Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/747,798 Pending US20220298259A1 (en) 2015-10-21 2022-05-18 Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer

Country Status (4)

Country Link
US (2) US20180312600A1 (ja)
EP (2) EP3365370A1 (ja)
JP (2) JP7078533B2 (ja)
WO (1) WO2017068164A1 (ja)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021127556A1 (en) * 2019-12-20 2021-06-24 Board Of Regents, The University Of Texas System Methods for treating cancer comprising low dose radiation
US11242404B2 (en) 2016-09-21 2022-02-08 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use
US11279766B2 (en) 2016-04-14 2022-03-22 Ose Immunotherapeutics Anti-SIRPa antibodies and their therapeutic applications
US11292850B2 (en) 2018-03-21 2022-04-05 ALX Oncology Inc. Antibodies against signal-regulatory protein α and methods of use
US11591390B2 (en) 2018-09-27 2023-02-28 Celgene Corporation SIRP-α binding proteins and methods of use thereof
US11713356B2 (en) 2017-10-13 2023-08-01 Ose Immunotherapeutics Modified bifunctional anti-human signal regulatory protein alpha (SIRPa) antibody and method of use thereof for treating cancer
US11884723B2 (en) 2018-03-13 2024-01-30 Ose Immunotherapeutics Use of anti-human SIRPa v1 antibodies and method for producing anti-SIRPa v1 antibodies

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3011097C (en) 2016-01-11 2022-12-20 Forty Seven, Inc. Humanized, mouse or chimeric anti-cd47 monoclonal antibodies
EP4070812A1 (en) * 2016-01-21 2022-10-12 The Board of Trustees of the Leland Stanford Junior University Treatment of cancer with combinations of immunoregulatory agents
WO2018107058A1 (en) 2016-12-09 2018-06-14 Alector Llc Anti-sirp-alpha antibodies and methods of use thereof
CN110799536B (zh) 2017-05-16 2024-04-26 拜奥迪斯私人有限公司 抗SIRPα抗体
PL3658589T3 (pl) 2017-07-26 2024-03-18 Forty Seven, Inc. Przeciwciała anty-sirp-alfa i powiązane sposoby
WO2019089653A1 (en) * 2017-10-30 2019-05-09 Apellis Pharmaceuticals, Inc. Treatment of disorders
SG11202005944VA (en) * 2017-12-22 2020-07-29 Atossa Therapeutics Inc Intraductal methods of treatment of breast disorders
PT3752190T (pt) 2018-02-12 2022-11-18 Forty Seven Inc Regime anticancerígeno usando anticorpos anti-cd47 e anti-cd20
AU2019272885A1 (en) 2018-05-25 2020-11-26 Alector Llc Anti-SIRPA antibodies and methods of use thereof
KR20210030267A (ko) * 2018-07-10 2021-03-17 고쿠리츠다이가쿠호진 고베다이가쿠 항 SIRPα 항체
JP2022535286A (ja) 2019-06-07 2022-08-05 エーエルエックス オンコロジー インコーポレイテッド 血清学的アッセイにおいてcd47に結合する薬物の干渉を低減するための方法及び試薬
IL293199A (en) 2019-11-27 2022-07-01 Alx Oncology Inc Combined therapies for cancer treatment
WO2021247430A1 (en) 2020-06-01 2021-12-09 ALX Oncology Inc. Combination therapies comprising a hypomethylation agent for treating cancer
EP4256336A1 (en) 2020-12-06 2023-10-11 ALX Oncology Inc. Multimers for reducing the interference of drugs that bind cd47 in serological assays
AU2022273727A1 (en) 2021-05-13 2023-11-09 ALX Oncology Inc. Combination therapies for treating cancer
WO2023020459A1 (zh) 2021-08-17 2023-02-23 杭州九源基因工程有限公司 靶向SIRPα的单克隆抗体及其用途
WO2023235754A1 (en) 2022-06-01 2023-12-07 ALX Oncology Inc. Combination therapies for treating urothelial carcinoma

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1048299A1 (en) 1999-04-28 2000-11-02 Faculteit der Geneeskunde van de Vrije Universiteit Method for inhibiting cell functioning for use in anti-inflammatory and anti-tumour therapies
EP2111869A1 (en) * 2008-04-23 2009-10-28 Stichting Sanquin Bloedvoorziening Compositions and methods to enhance the immune system
EP2995315B1 (en) 2009-05-15 2023-12-27 University Health Network Compositions and methods for treating hematologic cancers targeting the sirp alpha- cd47 interaction
US20140193408A1 (en) 2011-06-16 2014-07-10 Novartis Ag Soluble proteins for use as therapeutics
US20140242095A1 (en) 2011-10-19 2014-08-28 University Health Network Antibodies and antibody fragments targeting sirp-alpha and their use in treating hematologic cancers
US10570204B2 (en) * 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
EP2876167A1 (en) 2013-11-21 2015-05-27 Institut Gustave Roussy Microbiota composition, as a marker of responsiveness to chemotherapy, and use of microbial modulators (pre-,pro- or synbiotics) for improving the efficacy of a cancer treatment
EP3116544A4 (en) 2014-03-11 2017-08-23 The Board of Trustees of the Leland Stanford Junior University Anti sirp-alpha antibodies and bi-specific macrophage enhancing antibodies
EP3012271A1 (en) * 2014-10-24 2016-04-27 Effimune Method and compositions for inducing differentiation of myeloid derived suppressor cell to treat cancer and infectious diseases

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11279766B2 (en) 2016-04-14 2022-03-22 Ose Immunotherapeutics Anti-SIRPa antibodies and their therapeutic applications
US11242404B2 (en) 2016-09-21 2022-02-08 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use
US11401338B2 (en) 2016-09-21 2022-08-02 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use
US11713356B2 (en) 2017-10-13 2023-08-01 Ose Immunotherapeutics Modified bifunctional anti-human signal regulatory protein alpha (SIRPa) antibody and method of use thereof for treating cancer
US11884723B2 (en) 2018-03-13 2024-01-30 Ose Immunotherapeutics Use of anti-human SIRPa v1 antibodies and method for producing anti-SIRPa v1 antibodies
US11292850B2 (en) 2018-03-21 2022-04-05 ALX Oncology Inc. Antibodies against signal-regulatory protein α and methods of use
US11939393B2 (en) 2018-03-21 2024-03-26 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use
US11591390B2 (en) 2018-09-27 2023-02-28 Celgene Corporation SIRP-α binding proteins and methods of use thereof
WO2021127556A1 (en) * 2019-12-20 2021-06-24 Board Of Regents, The University Of Texas System Methods for treating cancer comprising low dose radiation

Also Published As

Publication number Publication date
US20220298259A1 (en) 2022-09-22
EP4186927A1 (en) 2023-05-31
JP2022040378A (ja) 2022-03-10
WO2017068164A1 (en) 2017-04-27
JP2018531274A (ja) 2018-10-25
EP3365370A1 (en) 2018-08-29
JP7078533B2 (ja) 2022-05-31

Similar Documents

Publication Publication Date Title
US20220298259A1 (en) Methods and compositions for modifying macrophage polarization into pro-inflammatory cells to treat cancer
JP2018531274A6 (ja) 炎症促進性細胞へのマクロファージ分極を改変して癌を治療するための方法及び組成物
EP3512548B1 (en) Antibody and checkpoint inhibitor combination therapy
US10501537B2 (en) Methods for treating cancer
US20230085710A1 (en) Novel polyinosinic - polycytidylic acid compositions
Fu et al. Combination foretinib and anti-PD-1 antibody immunotherapy for colorectal carcinoma
KR20200015469A (ko) 항-egfr/고친화성 nk-세포 조성물 및 척색종 치료 방법
AU2017345286A1 (en) Methods for promoting T cells response
US20230088070A1 (en) Use of il-1beta binding antibodies
EA037354B1 (ru) Лечение рака онколитическим вирусом в комбинации с ингибитором контрольных точек
WO2018039332A1 (en) Immunoswitch nanoparticles for reprogrammed t cell responses
WO2013187983A1 (en) Methods an compositions for treating or diagnosing melanoma
WO2015026831A1 (en) Compositons and methods for the treatment of radiation exposure
JP2021517908A (ja) トランスポーター阻害剤を含有する医薬品、医薬組成物及びその使用
Teng et al. Interferon gamma induces higher neutrophil extracellular traps leading to tumor-killing activity in microsatellite stable colorectal cancer
US20190177765A1 (en) Combination Therapies and Methods of Use Thereof
US20230406949A1 (en) Combinatorial immunotherapeutic methods and compositions for pancreatic ductal adenocarcinoma treatment
WO2018200473A1 (en) Detection and targeting of tumor-promoting neutrophils cross-reference to related applications
Yan et al. Cell targeting and immunostimulatory properties of a novel Fcγ-receptor-independent agonistic anti-CD40 antibody in rhesus macaques
Kudo-Saito et al. Targeting myeloid villains in the treatment with immune checkpoint inhibitors in gastrointestinal cancer
Cederberg Investigating the role of myeloid cells within the tumor microenvironment
Westerholm CD47 and TREM2 for macrophage-targeted cancer immunotherapy: Antibody validation and ex vivo efficacy in patient-derived explant
Pinho-Schwermann et al. Androgen receptor signaling blockade enhances NK cell-mediated killing of prostate cancer cells and sensitivity to NK cell checkpoint blockade
US20220025036A1 (en) Use of il-1beta binding antibodies
Indino IMPAIRMENT OF TOLL-LIKE RECEPTOR 9 AGONIST ANTI-TUMOR ACTIVITY BY ANTI-PD-1 ANTIBODY: ROLE OF MACROPHAGES

Legal Events

Date Code Title Description
AS Assignment

Owner name: OSE IMMUNOTHERAPEUTICS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:POIRIER, NICOLAS;VANHOVE, BERNARD;GAUTTIER, VANESSA;REEL/FRAME:046501/0541

Effective date: 20180720

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION